ArticlePDF AvailableLiterature Review

Role of epoxyeicosatrienoic acids in protecting the myocardium following ischemia/reperfusion injury

Authors:

Abstract and Figures

Cardiomyocyte injury following ischemia-reperfusion can lead to cell death and result in cardiac dysfunction. A wide range of cardioprotective factors have been studied to date, but only recently has the cardioprotective role of fatty acids, specifically arachidonic acid (AA), been investigated. This fatty acid can be found in the membranes of cells in an inactive state and can be released by phospholipases in response to several stimuli, such as ischemia. The metabolism of AA involves the cycloxygenase (COX) and lipoxygenase (LOX) pathways, as well as the less well characterized cytochrome P450 (CYP) monooxygenase pathway. Current research suggests important differences with respect to the cardiovascular actions of specific CYP mediated arachidonic acid metabolites. For example, CYP mediated hydroxylation of AA produces 20-hydroxyeicosatetraenoic acid (20-HETE) which has detrimental effects in the heart during ischemia, pro-inflammatory effects during reperfusion and potent vasoconstrictor effects in the coronary circulation. Conversely, epoxidation of AA by CYP enzymes generates 5,6-, 8,9-, 11,12- and 14,15-epoxyeicosatrienoic acids (EETs) that have been shown to reduce ischemia-reperfusion injury, have potent anti-inflammatory effects within the vasculature, and are potent vasodilators in the coronary circulation. This review aims to provide an overview of current data on the role of these CYP pathways in the heart with an emphasis on their involvement as mediators of ischemia-reperfusion injury. A better understanding of these relationships will facilitate identification of novel targets for the prevention and/or treatment of ischemic heart disease, a major worldwide public health problem.
Content may be subject to copyright.
Role of epoxyeicosatrienoic acids in protecting the myocardium
following ischemia/reperfusion injury
John M. Seuberta,*, Darryl C. Zeldinb, Kasem Nithipatikomc, and Garrett J. Grossc
a Faculty of Pharmacy and Pharmaceutical Sciences, 3126 Dentistry/Pharmacy Centre, University of Alberta,
Edmonton, AB, Canada T6G 2N8
b Division of Intramural Research, NIEHS/NIH, Research Triangle Park, NC 27709, USA
c Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
Abstract
Cardiomyocyte injury following ischemia–reperfusion can lead to cell death and result in cardiac
dysfunction. A wide range of cardioprotective factors have been studied to date, but only recently
has the cardioprotective role of fatty acids, specifically arachidonic acid (AA), been investigated.
This fatty acid can be found in the membranes of cells in an inactive state and can be released by
phospholipases in response to several stimuli, such as ischemia. The metabolism of AA involves the
cycloxygenase (COX) and lipoxygenase (LOX) pathways, as well as the less well characterized
cytochrome P450 (CYP) monooxygenase pathway. Current research suggests important differences
with respect to the cardiovascular actions of specific CYP mediated arachidonic acid metabolites.
For example, CYP mediated hydroxylation of AA produces 20-hydroxyeicosatetraenoic acid (20-
HETE) which has detrimental effects in the heart during ischemia, pro-inflammatory effects during
reperfusion and potent vasoconstrictor effects in the coronary circulation. Conversely, epoxidation
of AA by CYP enzymes generates 5,6-, 8,9-, 11,12- and 14,15-epoxyeicosatrienoic acids (EETs) that
have been shown to reduce ischemia–reperfusion injury, have potent anti-inflammatory effects
within the vasculature, and are potent vasodilators in the coronary circulation. This review aims to
provide an overview of current data on the role of these CYP pathways in the heart with an emphasis
on their involvement as mediators of ischemia–reperfusion injury. A better understanding of these
relationships will facilitate identification of novel targets for the prevention and/or treatment of
ischemic heart disease, a major worldwide public health problem.
Keywords
Arachidonic acid; Cytochrome P450; Eicosanoids; Ischemia reperfusion injury; Cardioprotection
1. Introduction
Heart disease and stroke are major causes of illness, disability and death in Western societies,
and impose a great burden to national health care systems [1-6]. For example, cardiovascular
disease (CVD) accounted for the death of approximately 76,500 Canadians in 2002 [2,4] and
over 910,000 individuals in the US [6]. As the population ages and co-morbidities, such as
obesity and diabetes become more prevalent, both the human cost and economic burden of
CVD will likely increase. Acute myocardial infarction (AMI) continues to be a leading cause
of death worldwide [7,8]. Myocardial infarction occurs when ischemia exceeds a critical
threshold and overwhelms cellular repair mechanisms that are designed to maintain normal
operating function and homeostasis. Ischemia at this critical threshold level results in
* Corresponding author. Tel.: +1 780 492 0007; fax: +1 780 492 1217. E-mail address:jseubert@pharmacy.ualberta.ca (J.M. Seubert).
NIH Public Access
Author Manuscript
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
Published in final edited form as:
Prostaglandins Other Lipid Mediat. 2007 January ; 82(1-4): 50–59.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
irreversible myocardial cell damage or death. Such injury contributes to the pathogenesis of
heart failure (HF), AMI and sudden death [9]. Advances in early reperfusion therapy, such as
thromobolytic drugs, coronary angioplasty or bypass graft surgery, have reduced morbidity,
HF and infarct-associated ventricular arrhythmias. Preconditioning (PC) is another powerful
cardioprotective strategy that renders the heart resistant to injury [10]. Brief, non-detrimental
episodes of ischemia or pharmacological mimetics given prior to a prolonged ischemic event
can initiate signaling events that protect the myocardium [10]. Unfortunately, both early
reperfusion therapy and cardioprotective drugs given prior to ischemia have limited clinical
utility as patients typically present after the onset of ischemia and/or are unable to reach medical
facilities [11,12]. In light of the increasing incidence and prevalence of HF after AMI [1,3,4],
there is a profound need for a better understanding of the underlying pathophysiology and a
need for development of strategies to protect the myocardium from ischemic-reperfusion
injury. Thus, novel therapeutic strategies are required in order to prevent the adverse
consequences and impact of CVD.
2. Arachidonic acid, CYP epoxygenases and soluble epoxide hydrolase
Arachidonic acid (AA), a polyunsaturated fatty acid normally found esterified to cell
membrane glycerophospholipids, can be released by phospholipases in response to several
stimuli, such as ischemia [13]. Free AA is then available for metabolism by prostaglandin H2
synthases, lipoxygenases and cytochrome P450 monooxygenases to generate numerous
metabolites, collectively termed eicosanoids [14,15]. CYP epoxygenases metabolize AA to
four regioisomeric epoxyeicosatrienoic acids (5,6-, 8,9-, 11,12- and 14,15-EETs), all of which
are biologically active (Fig. 1) [16,17]. The actions of EETs are terminated by conversion to
the corresponding and less biologically active dihydroxyeicosatrienoic acids (DHETs) by
epoxide hydrolases [13]. Two major epoxide hydrolases are found in mammalian tissues, the
microsomal epoxide hydrolase (mEH) and the soluble epoxide hydrolase (sEH or EPHX2)
[14]. Previous work has demonstrated that sEH is the main enzyme involved in the in vivo
hydrolysis of the EETs [15,17]. The majority of endogenous EETs (>85%) are esterified to
membrane glycerophospholipids, particularly phosphatidylcholine and phosphatidylinositol,
where they are generally considered inactive until their release [17]. In this regard, ischemia
has been shown to activate cytosolic phospholipase A2 leading to the release of bioactive
eicosanoids from glycerophospholipids [18].
EETs are important components of many intracellular signaling pathways in both cardiac and
extracardiac tissues. For example, EETs activate Ca2+-sensitive K+ channels (BKCa) in
vascular smooth muscle cells resulting in hyperpolarization of the resting membrane potential
and vasodilation of the coronary circulation [19-21]. This effect is diminished upon hydrolysis
of EETs to DHETs by sEH [22]. Other studies have shown that EETs display anti-
inflammatory, thrombolytic and angiogenic properties within the vasculature [23-25]. In
endothelial cells, EETs activate mitogen activated protein kinase (MAPK) and
phosphatidylinositol-3 kinase (PI3K)-Akt signaling pathways [25], increase intracellular
cAMP levels [24], upregulate expression of nitric oxide synthase [26] and protect against
hypoxia-reoxygenation injury [27]. In general, the effects of DHETs on these pathways are
less pronounced [23,24]. Within the heart, EETs activate cardiac ATP-sensitive K+ (KATP)
channels [28-31], enhance L-type calcium currents [32,33] and improve postischemic recovery
of left ventricular function [31,34-36]. Thus, alteration in the production and/or elimination of
EETs may affect steady-state cellular levels of these bioactive eicosanoids in vivo and could
potentially influence cardiac function (Fig. 2).
Arachidonic acid can be metabolized by CYP epoxygenases and CYP ω-hydroxylases to
products that have vastly different physiologic effects. For example, EETs have potent
vasodilatory properties [37] and 20-hydroxyeicosatetraenoic acid (20-HETE) has potent
Seubert et al. Page 2
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
vasoconstrictive effects [38]. Therefore, changes in the expression and/or activity of specific
CYP epoxygenase and hydroxylase enzymes can alter the delicate balance between EETs and
20-HETE. For instance, recent data has demonstrated that inhibition of CYP ω-hydroxylases
results in reduction of infarct size in rats and dogs following ischemic injury, which suggests
that 20-HETE has detrimental effects in the heart [39,40]. These investigators originally
showed that 20-HETE was released into coronary venous blood in high concentrations at the
end of 60 min of ischemia and throughout 3 h of reperfusion. Furthermore, the non-selective
CYP inhibitor, miconazole (MIC) and the more selective inhibitors, 17-octadecanoic acid (17-
ODYA) and N-methylsulfonyl-12,12-dibromododec-11-enamide (DDMS), all reduced 20-
HETE release from the ischemic/reperfused heart and produced marked reductions in
myocardial infarct size (expressed as a percent of the area at risk) from 19.6 ± 1.7% to 8.4 ±
2.5% (MIC), 5.9 ± 2.2% (17-ODYA) and 10.8 ± 1.8% (DDMS), respectively [39,40].
Conversely, exogenous administration of 20-HETE significantly increased infarct size to 26.9
± 1.9%. These investigators also showed that at least three isoforms of the ω-hydroxylases
were present in canine heart tissue – CYP4A1, CYP4A2 and CYP4F – all of which were
markedly inhibited by incubation with 17-ODYA. Reductions in infarct size were also
observed by this same group in rat hearts in which MIC,17-ODYA and DDMS were
adminstered prior to ischemia or 5 min prior to reperfusion [40] These results suggest that the
major effect of inhibiting ω-hydroxylase in rats occurs during reperfusion and is associated
with in a decrease in “reperfusion injury” More recently, this same group demonstrated that
inhibition of ω-hydroxylases in dog hearts with DDMS reduced infarct size to a degree equal
to that of ischemic PC, the “gold standard” in cardioprotection research. These same
investigators found that the administration of the 20-HETE antagonist, 20-HEDE, reduced
infarct size similar to that observed with ischemic PC and DDMS. Interestingly, the
combination of ischemic PC and DDMS produced a significantly greater reduction in infarct
size than either intervention alone. Together, the results suggest that these two treatments may
be acting via different signaling pathways and may be a potent combination in alleviating the
sequelae of ischemia and/or reperfusion injury in a clinical setting, such as coronary artery
bypass graft surgery.
Transgenic hearts from mice overexpressing human CYP2J2 have improved postischemic
functional recovery as evidenced by studies using Langendorff perfused hearts subjected to
ischemia/reperfusion injury [31]. Perfusion with the selective P450 epoxygenase inhibitor N-
methylsulphonyl-6-(2-proparglyloxyphenyl)hexanamide (MS-PPOH) for 20 min prior to
ischemia resulted in reduction of postischemic LVDP recovery in wild type hearts and
abolished the improved postischemic LVDP recovery in CYP2J2 transgenic hearts. These data
provided evidence that the cardioprotective effects of CYP2J2 overexpression involved P450
epoxygenase metabolites. Moreover, these data suggest an important role for endogenous P450
epoxygenases in postischemic functional recovery. Mechanistic studies by these investigators
[31] suggested that enhanced activation of ATP-sensitive K+ channels (KATP) were involved
in the improved recovery observed in the CYP2J2 transgenic mice. In addition, CYP2J2
overexpressing mice exhibited increased expression of phospho-p42/p44 mitogen-activated
protein kinase (MAPK) following ischemia. The addition of the p42/p44 MAPK kinase (MEK)
inhibitor PD98059 during reperfusion abolished the cardioprotective effects observed in the
CYP2J2 mice. Together, these data suggest that CYP2J2-derived metabolites are
cardioprotective following ischemia and the mechanism for this cardioprotection involves
activation of KATP channels and p42/p44 MAPK.
Many CYP inhibitors lack isoform specificity and also may have effects on other signaling
pathways. Reduction in infarct size observed in rat and rabbit hearts following treatment with
non-specific CYP inhibitors has been attributed to suppression of CYP-dependent ROS
production [41]. CYP2C and CYP2J isozymes are the predominant AA epoxygenases in the
cardiovascular system. While CYP2C9 has been shown to generate ROS in coronary arteries
Seubert et al. Page 3
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
[42], CYP2J2 is not a relevant source of ROS [27,42]. These studies highlight the complexity
of the CYP enzyme system, emphasize the role of different CYP metabolites in
cardioprotection and suggest caution in the interpretation of results when using non-selective
CYP inhibitors. Recent epidemiologic data suggests an association between single nucleotide
polymorphisms in the genes encoding CYP2J2, CYP2C8, CYP2C9 and EPHX2 and
cardiovascular disease risk in humans supporting the functional relevance of the CYP
epoxygenase pathway in the heart [43-47].
3. Ischemic injury and cardioprotection
Ischemic heart disease is an underlying cause of most AMIs, congestive HF, arrhythmias and
sudden cardiac death. Myocardial ischemia is characterized by inadequate blood flow to the
heart resulting in limited glucose, oxygen and delayed metabolic by-product removal.
Ultimately, ischemic events result in cellular death and myocyte loss which is the primary
pathology behind many CVDs. Myocytes are not easily replaced, although stem cell therapy
shows great initial promise in overcoming this problem. Nevertheless, preserving the viability
of ischemic myocardium is the major goal for cardioprotection. Cardioprotective mechanisms
modulate cell metabolism or signaling pathways directly or via posttranslational modification
of key proteins, as well initiate new transcription and translation [10,12]. A diverse spectrum
of signals converge onto a few common end effectors that maintain membrane integrity and
inhibit cell death [48]. Mitochondria are one of these important convergence points, due to
their critical function in cell survival and death; notably, ATP production and apoptosis [10,
12,48]. Key mitochondrial proteins, such as several potassium (K+) channels [49-53] and the
mitochondrial permeability transition pore (mPTP) [48,54-56], act as effectors integrating
these upstream signals into cardioprotective responses. The mechanisms by which CYP
metabolites alter these important effectors are not well defined and require further
experimentation.
3.1. K+ channels and cardioprotection
K+ channels are membrane proteins involved in many physiological processes, such as
regulation of heart rate, muscle contraction and cell volume [57]. Two pharmacologically
distinct ATP-sensitive potassium channels (KATP) have been identified in cardiomyocytes,
sarcolemmal KATP (Sarc KATP) and mitochondrial KATP (mito KATP) [58]. sarc KATP is
activated during cardiac ischemia when cytoplasmic ATP is depleted and this affects membrane
excitability. Activation leads to shortening of the cardiac action potential and reduced
intracellular calcium overload [59,60]. Although there are no selective sarc KATP openers, a
number of drugs known to open this channel have been shown to produce beneficial effects in
the myocardium in animal models of ischemia/reperfusion injury, and several non-selective
inhibitors of sarc KATP (such as glibenclamide) block ischemic PC [59,60]. In spite of these
findings, the strongest evidence for a role of sarc KATP channels in ischemic PC has been
demonstrated in the KiR 6.2 knockout mouse where ischemic PC could not be produced [60].
The EETs have been shown to be potent openers of sarc KATP by reducing channel sensitivity
to ATP in isolated rat myocytes using the inside-out patch clamp technique; however, the exact
site on the channel that interacts with EETs remains unknown [30,61]. These authors also
showed that membrane hyperpolarization occurred in isolated rat myocytes by 11,12-EET
addition, an effect blocked by glibenclamide, the non-selective KATP channel inhibitor. A role
for the sarc KATP channel in mediating the cardioprotective effect of inhibiting ω-hydroxylase
at the time of reperfusion in the rat heart was recently described [34]. These investigators
showed that the cardioprotective effect resulting from administering MIC and 17-ODYA just
prior to reperfusion in rats was completely abolished by HMR 1098, a selective inhibitor of
the sarc KATP channel. In contrast, the selective mito KATP channel inhibitor, 5-
hydroxydecanoic acid (5-HD), had no effect. Although it can be assumed that a shift in the 20-
Seubert et al. Page 4
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
HETE/EET balance in favor of the EETs is responsible for the cardioprotective effect of
inhibiting 20-HETE synthesis, it remains unknown if the cardioprotective effect of the ω-
hydroxylase inhibitors is the result of the action of the EETs on the sarc KATP channel in the
myocardium.
In contrast to the sarc KATP channel, the precise molecular composition of the mito KATP
channel remains elusive. However, recent studies suggest it is part of a multiprotein complex
including the adenine nucleotide transporter (ANT) and succinate dehydrogenase (SDH)
[62]. Importantly, pharmacologic data indicate that selective activation of mito KATP confers
cardioprotection following ischemia and that this channel is the major one mediating ischemic
PC [59,60,63,64]. While the precise pathways by which mito KATP activation confers
cardioprotection remain unknown, potentially beneficial consequences of opening mito
KATP include partial depolarization of the intramitochondrial membrane, transient swelling of
the intramitochondrial space, enhanced respiration via the electron transport chain, reduced
mitochondrial calcium overload, and altered production of reactive oxygen species [59,60,
64]. Cardioprotective effects of CYP2J2 overexpression involve activation of mito KATP
channels [31]. Increased flavoprotein fluorescence (a marker of mitochondrial redox status)
[63] in CYP2J2 Tr cardiomyocytes is consistent with enhanced mito KATP activation in the
presence of CYP2J2 overexpression [31]. Moreover, treatment of wild type cardiomyocytes
with physiologically relevant concenrations of EETs increased flavoprotein fluorescence
[31].
Ca2+-activated K+ (KCa) channels include large conductance (BKCa), voltage-sensitive K+
selective proteins expressed in various tissues including heart mitochondria [53]. KCa channels
can be activated by elevations in intracellular Ca2+ and membrane depolarization [65,66]. CYP
epoxygenase derived EETs are known activators of BKCa channels in vascular smooth muscle
[20,67], whereas, CYP ω-hydroxylases derived HETEs are known inhibitors in vascular
smooth muscle [38,68,69]. Recent evidence suggests that newly identified KCa channels in
cardiac mitochondria (mito KCa) [53,70] are important mediators of cardioprotection. It is
proposed that these mitochondrial K+ channels work in concert with mito KATP and other
mitochondrial proteins in response to ischemia [49]. Activation of K+ channels by kinases,
such as PKC or PKA, and other unknown signals is predicted to increase mitochondrial K+
uptake and in turn reduce Ca2+ overload in cardiomyocytes. The cardioprotective mechanisms
associated with opening of these channels include a mild uncoupling, depolarization of the
intramitochondrial membrane, transient swelling of the intramitochondrial space, enhanced
respiration via the electron transport chain and altered production of reactive oxygen species
[49,53,59,60,64,70].
3.2. Mitochondrial permeability transition pore
mPTP is a protein complex on the inner mitochondrial membrane which includes ANT,
cyclophilin D and the voltage-dependent anion channel (VDAC) [48,49,71]. mPTP remains
closed under normal physiological conditions but opens under cellular stress, such as during
reperfusion following an ischemic event [48,49,54]. Opening allows free passage of molecules
>1.5 kDa which initiate adverse effects, like large osmotic pressure changes and uncoupling
of oxidative phosphorylation, that ultimately result in cell death [48,72]. Mitochondrial Ca2+
overload, oxidative stress, adenine nucleotide depletion and mitochondrial depolarization
contribute to the pore opening. Inhibition of prolonged mPTP opening (high-conductance)
during reperfusion with cyclosporine-A (CsA) or sanglifehrin-A (SfA) can reduce
cardiomyocyte injury [73-75]. Conversely, evidence suggests that the transient (low-
conductance) opening of mPTP during ischemic PC plays a role in cell survival [54]. Cell
culture experiments suggest that mito KATP openers, such as diazoxide, trigger mPTP opening
which can be blocked by CsA or 5HD [54]. Recent evidence demonstrates that multiple
Seubert et al. Page 5
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
cardioprotective kinases, PKA, PKB/Akt and PKC can converge upon glycogen synthase
kinase-3β (GSK-3β), which will induce inhibition of the mPTP [76]. While it is very likely
that signaling pathways involved in EET-mediated cardioprotection target the mitochondria,
it is unknown whether the response involves transient opening of mPTP prior to ischemia or
prevents prolonged opening during reperfusion. EETs have been shown to activate sarc
KATP [30,61] and mito KATP channels [31] in the heart, and KCa channels [20,67], in vascular
smooth muscle cells [38,68,69]. Increased flavoprotein fluorescence in CYP2J2 Tr
cardiomyocytes and wild type cardiomyocytes treated with EETs [31], strongly suggest
convergence of a cardioprotective signal onto the mitochondria. It is expected that EETs will
alter the flavin nucleotide fluorescence and intracellular [Ca2+] within the myocytes and most
likely alter mPTP opening. It is unknown whether EETs work solely via mito KATP channels;
however, it is likely they work together with other proteins, such as mito KCa channels. Future
experiments will help determine whether EETs influence the transient opening or closing of
mPTP, as well how they influence intramitochondrial [Ca2+] levels.
4. Cardioprotective signaling pathways
There is considerable controversy regarding the role of cytochrome P450s in the heart, notably
the beneficial versus the detrimental effects of arachidonic acid metabolites [31,34,35,39,40,
77,78]. We have demonstrated that EETs play a significant role in the improved postischemic
functional recovery in isolated mouse hearts overexpressing CYP2J2 [31,34,35,57,78]. Recent
results suggest potential cardioprotective mechanisms and indicate that KATP channels, p42/
p44-MAPK and PI3K are involved. However, as previously discussed EETs are known to
activate a number of signaling elements and ion channels, therefore, further work needs to be
done to determine the important protective mediators involved in this novel cardioprotective
pathway. Preliminary data suggested that EETs might only partially activate [31] the mito
KATP channel and could be working synergistically with other cardioprotective sites, such as
the sarc KATP channel or the mito KCa channel. Additional evidence has demonstrated an
important role for PKA and PKC in the cardioprotective pathway involving K+ channels [79,
80,81]. Further experiments will help determine if EET-mediated cardioprotection involves
early activation or co-localization of PKA or PKC to the mitochondria.
5. Conclusion
There are numerous reasons for investigating the role of this novel endogenous pathway in
myocardial function and ischemic injury. First, CYP-derived metabolites of arachidonic acid
play critical roles in modulating fundamental biological processes [68,82]. Second,
environmental or genetic factors that alter P450 expression and/or function lead to changes in
the production of bioactive eicosanoids [68,82]. Such effects can influence cell and organ
function in either an adverse or beneficial manner. Third, CYP isozymes expressed in the heart,
notably CYP2J2 [36,83], generate EETs, which have been shown to be cardioprotective [31,
36,83]. In contrast, other CYP isozymes (CYP2C and CYP4A) generate products, which are
thought to be detrimental to the heart, such as ROS and 20-HETE [39-42]. Traditionally,
investigation into the role of CYP isozymes has focused on hepatic and renal drug metabolism
and function. There is little known about the importance of this endogenous system within the
heart even though the heart contains significant levels of functionally active CYP. Finally, and
most importantly, recent human epidemiological evidence has identified associations between
CYP2J2 polymorphisms and CAD, and EPHX2 polymorphisms and CHD [43,45,46,84]. These
findings provide strong evidence supporting the notion that EETs play an important role in
postischemic functional recovery and perhaps infarct size reduction [31,33,36,83]. Recent
studies have only begun to address the underlying mechanisms of EET-mediated
cardioprotection and highlight the potential for this novel endogenous system as a therapeutic
target for CVD. Ultimately, understanding the basic cellular mechanisms of EET-mediated
Seubert et al. Page 6
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
cardioprotection will enhance our knowledge of this important phenomenon and will lead to
the development of novel therapeutics for the treatment of cardiovascular diseases. Targeting
CYP2J2, ω-hydroxylases or sEH, either through pharmacological or gene therapy methods,
represents a novel therapeutic approach to the management of ischemic heart disease in
humans.
Abbreviations
AA, arachidonic acid; AMI, acute myocardial infarction; ANT, adenine nucleotide transporter;
BKCa, large conductance calcium activated potassium channels; CYP, cytochrome P450
monooxygenase; CVD, cardiovascular disease; DHET, dihydroepoxyeicosatrienoic acid;
EETs, epoxyeicosatrienoic acid; GSK-3β, glycogen synthase kinase-3β; 20-HETE, 20-
hydroxyeicosatetraenoic acid; HF, heart failure; IPC, ischemic preconditioning; IR, ischemic-
reperfusion; KATP, ATP-sensitive potassium channel; KCa, calcium-activated potassium
channel; LVDP, left ventricular developed pressure; MAPK, mitogen activated protein kinase;
mEH, microsomal epoxide hydrolase; mito KATP, mitochondrial ATP-sensitive potassium
channel; mPTP, mitochondrial permeability transition pore; MS-PPOH, N-methylsulphonyl-6-
(2-proparglyloxyphenyl) hexanamide; PKA, protein kinase A; PKC, protein kinase C; PI3K,
phosphatidylinositol-3 kinase; sarc KATP, sarcolemmal ATP-sensitive potassium channel;
sEH/Ephx2, soluble epoxide hydrolase; SDH, succinate dehydrogenase; VDAC, voltage-
dependent anion channel.
References
1. Health Canada. Economic burden of illness in Canada. 1998.
2. Heart and Stroke Foundation of Canada. The growing burden of heart disease and stroke in Canada.
2003.
3. Manuel DG, Leung M, Nguyen K, Tanuseputro P, Johansen H. Burden of cardiovascular disease in
Canada. Can J Cardiol 2003;19:997–1004. [PubMed: 12915926]
4. Manuel DG, Luo W, Ugnat AM, Mao Y. Cause-deleted health-adjusted life expectancy of Canadians
with selected chronic conditions. Chronic Dis Can 2003;24:108–15. [PubMed: 14733760]
5. Statistics Canada. Causes of death, 2002. 2004.
6. Thom T, Haase N, Rosamond W, et al. Heart disease and stroke statistics—2006 update: a report from
the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Circulation
2006;113:e85–e151. [PubMed: 16407573]
7. Tu JV, Austin PC, Filate WA, Johansen HL, et al. Outcomes of acute myocardial infarction in Canada.
Can J Cardiol 2003;19:893–901. [PubMed: 12876609]
8. Tu JV, Cameron C. Impact of an acute myocardial infarction report card in Ontario, Canada. Int J Qual
Health Care 2003;15:131–7. [PubMed: 12705706]
9. Bolli R, Becker L, Gross G, Mentzer R Jr, Balshaw D, Lathrop DA. Myocardial protection at a
crossroads: the need for translation into clinical therapy. Circ Res 2004;95:125–34. [PubMed:
15271864]
10. Yellon DM, Downey JM. Preconditioning the myocardium: from cellular physiology to clinical
cardiology. Physiol Rev 2003;83:1113–51. [PubMed: 14506302]
11. Cannon R III. Mechanisms, management and future directions for reperfusion injury after myocardial
infarction. Nat Clin Pract 2005;2:88–94.
12. Murphy E. Primary and secondary signaling pathways in early preconditioning that converge on the
mitochondria to produce cardioprotection. Circ Res 2004;94:7–16. [PubMed: 14715531]
13. Morisseau C, Hammock BD. Epoxide hydrolases: mechanisms, inhibitor designs, and biological
roles. Annu Rev Pharmacol Toxicol. 2004
14. Fang X, Weintraub NL, McCaw RB, et al. Effect of soluble epoxide hydrolase inhibition on
epoxyeicosatrienoic acid metabolism in human blood vessels. Am J Physiol Heart Circ Physiol
2004;287:H2412–20. [PubMed: 15284062]
Seubert et al. Page 7
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
15. Zeldin DC, Kobayashi J, Falck JR, et al. Regio- and enantiofacial selectivity of epoxyeicosatrienoic
acid hydration by cytosolic epoxide hydrolase. J Biol Chem 1993;268:6402–7. [PubMed: 8454612]
16. Spector AA, Fang X, Snyder GD, Weintraub NL. Epoxyeicosatrienoic acids (EETs): metabolism and
biochemical function. Prog Lipid Res 2004;43:55–90. [PubMed: 14636671]
17. Zeldin DC, DuBois RN, Falck JR, Capdevila JH. Molecular cloning, expression and characterization
of an endogenous human cytochrome p450 arachidonic acid epoxygenase isoform. Arch Biochem
Biophys 1995;322:76–86. [PubMed: 7574697]
18. Saluja I, Song D, O'Regan MH, Phillis JW. Role of phospholipase a2 in the release of free fatty acids
during ischemia–reperfusion in the rat cerebral cortex. Neurosci Lett 1997;233:97–100. [PubMed:
9350841]
19. Campbell WB, Gebremedhin D, Pratt PF, Harder DR. Identification of epoxyeicosatrienoic acids as
endothelium-derived hyperpolarizing factors. Circ Res 1996;78:415–23. [PubMed: 8593700]
20. Fisslthaler B, Popp R, Kiss L, et al. Cytochrome p450 2c is an edhf synthase in coronary arteries.
Nature 1999;401:493–7. [PubMed: 10519554]
21. Fleming I. Cytochrome p450 epoxygenases as edhf synthase(s). Pharmacol Res 2004;49:525–33.
[PubMed: 15026030]
22. Fang X, Kaduce TL, Weintraub NL, et al. Pathways of epoxyeicosatrienoic acid metabolism in
endothelial cells. Implications for the vascular effects of soluble epoxide hydrolase inhibition. J Biol
Chem 2001;276:14867–74. [PubMed: 11278979]
23. Node K, Huo Y, Ruan X, et al. Anti-inflammatory properties of cytochrome p450 epoxygenase-
derived eicosanoids. Science 1999;285:1276–9. [PubMed: 10455056]
24. Node K, Ruan XL, Dai J, et al. Activation of gas mediates induction of tissue-type plasminogen
activator gene transcription by epoxyeicosatrienoic acids. J Biol Chem 2001;276:15983–9. [PubMed:
11279071]
25. Wang Y, Wei X, Xiao X, et al. Arachidonic acid epoxygenase metabolites stimulate endothelial cell
growth and angiogenesis via mitogen-activated protein kinase and phosphatidylinositol 3-kinase/Akt
signaling pathways. J Pharmacol Exp Ther 2005;314:522–32. [PubMed: 15840765]
26. Wang H, Lin L, Jiang J, et al. Up-regulation of endothelial nitric-oxide synthase by endothelium-
derived hyperpolarizing factor involves mitogen-activated protein kinase and protein kinase c
signaling pathways. J Pharmacol Exp Ther 2003;307:753–64. [PubMed: 12975498]
27. Yang B, Graham L, Dikalov S, et al. Overexpression of cytochrome p450 cyp2j2 protects against
hypoxia-reoxygenation injury in cultured bovine aortic endothelial cells. Mol Pharmacol
2001;60:310–20. [PubMed: 11455018]
28. Lee HC, Lu T, Weintraub NL, VanRollins M, Spector AA, Shibata EF. Effects of epoxyeicosatrienoic
acids on the cardiac sodium channels in isolated rat ventricular myocytes. J Physiol 1999;519(Pt 1):
153–68. [PubMed: 10432346]
29. Lu T, Hoshi T, Weintraub NL, Spector AA, Lee HC. Activation of ATP-sensitive k(+) channels by
epoxyeicosatrienoic acids in rat cardiac ventricular myocytes. J Physiol 2001;537:811–27. [PubMed:
11744757]
30. Lu T, VanRollins M, Lee HC. Stereospecific activation of cardiac ATP-sensitive k(+) channels by
epoxyeicosatrienoic acids: a structural determinant study. Mol Pharmacol 2002;62:1076–83.
[PubMed: 12391270]
31. Seubert J, Yang B, Bradbury JA, et al. Enhanced postischemic functional recovery in cyp2j2
transgenic hearts involves mitochondrial ATP-sensitive K+ channels and p42/p44 mapk pathway.
Circ Res 2004;95:506–14. [PubMed: 15256482]
32. Xiao YF, Huang L, Morgan JP. Cytochrome p450: a novel system modulating Ca2+ channels and
contraction in mammalian heart cells. J Physiol 1998;508:777–92. [PubMed: 9518732]
33. Xiao YF, Ke Q, Seubert JM, et al. Enhancement of cardiac l-type Ca2+ currents in transgenic mice
with cardiac-specific overexpression of cyp2j2. Mol Pharmacol 2004;66:1607–16. [PubMed:
15361551]
34. Nithipatikom K, Endsley MP, Moore JM, et al. Effects of selective inhibition of cytochrome p-450
omega-hydroxylases and ischemic preconditioning in myocardial protection. Am J Physiol Heart
Circ Physiol 2006;290:H500–5. [PubMed: 16214838]
Seubert et al. Page 8
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
35. Nithipatikom K, Moore JM, Isbell MA, Falck JR, Gross GJ. Epoxyeicosatrienoic acids (EETs) in
cardioprotection: ischemic versus reperfusion injury. Am J Physiol Heart Circ Physiol. 2006
36. Wu S, Chen W, Murphy E, et al. Molecular cloning, expression, and functional significance of a
cytochrome p450 highly expressed in rat heart myocytes. J Biol Chem 1997;272:12551–9. [PubMed:
9139707]
37. Medhora M, Narayanan J, Harder D. Dual regulation of the cerebral microvasculature by
epoxyeicosatrienoic acids. Trends Cardiovasc Med 2001;11:38–42. [PubMed: 11413051]
38. Roman RJ, Maier KG, Sun CW, Harder DR, Alonso-Galicia M. Renal and cardiovascular actions of
20-hydroxyeicosatetraenoic acid and epoxyeicosatrienoic acids. Clin Exp Pharmacol Physiol
2000;27:855–65. [PubMed: 11071299]
39. Gross ER, Nithipatikom K, Hsu AK, et al. Cytochrome p450 omega-hydroxylase inhibition reduces
infarct size during reperfusion via the sarcolemmal kATP channel. J Mol Cell Cardiol 2004;37:1245–
9. [PubMed: 15572055]
40. Nithipatikom K, Gross ER, Endsley MP, et al. Inhibition of cytochrome p450omega-hydroxylase: a
novel endogenous cardioprotective pathway. Circ Res 2004;95:e65–71. [PubMed: 15388642]
41. Granville DJ, Tashakkor B, Takeuchi C, et al. Reduction of ischemia and reperfusion-induced
myocardial damage by cytochrome p450 inhibitors. Proc Natl Acad Sci USA 2004;101:1321–6.
[PubMed: 14734800]
42. Fleming I, Michaelis UR, Bredenkotter D, et al. Endothelium-derived hyperpolarizing factor synthase
(cytochrome p450 2c9) is a functionally significant source of reactive oxygen species in coronary
arteries. Circ Res 2001;88:44–51. [PubMed: 11139472]
43. Fornage M, Boerwinkle E, Doris PA, Jacobs D, Liu K, Wong ND. Polymorphism of the soluble
epoxide hydrolase is associated with coronary artery calcification in African–American subjects. The
coronary artery risk development in young adults (cardia) study. Circulation. 2004
44. Fornage M, Lee CR, Doris PA, et al. The soluble epoxide hydrolase gene harbors sequence variation
associated with susceptibility to and protection from incident ischemic stroke. Hum Mol Genet
2005;14:2829–37. [PubMed: 16115816]
45. Przybyla-Zawislak BD, Srivastava PK, Vazquez-Matias J, et al. Polymorphisms in human soluble
epoxide hydrolase. Mol Pharmacol 2003;64:482–90. [PubMed: 12869654]
46. Spiecker M, Darius H, Hankeln T, et al. Risk of coronary artery disease associated with polymorphism
of the cytochrome p450 epoxygenase cyp2j2. Circulation 2004;110:2132–6. [PubMed: 15466638]
47. Yasar U, Bennet AM, Eliasson E, et al. Allelic variants of cytochromes p450 2c modify the risk for
acute myocardial infarction. Pharmacogenetics 2003;13:715–20. [PubMed: 14646690]
48. Halestrap AP, Clarke SJ, Javadov SA. Mitochondrial permeability transition pore opening during
myocardial reperfusion—a target for cardioprotection. Cardiovasc Res 2004;61:372–85. [PubMed:
14962470]
49. Hanley PJ, Daut J. K(ATP) channels and preconditioning: a re-examination of the role of
mitochondrial k(ATP) channels and an overview of alternative mechanisms. J Mol Cell Cardiol
2005;39:17–50. [PubMed: 15907927]
50. Liu Y, Sato T, Seharaseyon J, Szewczyk A, O'Rourke B, Marban E. Mitochondrial ATP-dependent
potassium channels. Viable candidate effectors of ischemic preconditioning. Ann NY Acad Sci
1999;874:27–37. [PubMed: 10415518]
51. O'Rourke B. Evidence for mitochondrial K+ channels and their role in cardioprotection. Circ Res
2004;94:420–32. [PubMed: 15001541]
52. Wang X, Yin C, Xi L, Kukreja RC. Opening of Ca2+-activated K+ channels triggers early and delayed
preconditioning against i/r injury independent of nos in mice. Am J Physiol Heart Circ Physiol
2004;287:H2070–7. [PubMed: 15217801]
53. Xu W, Liu Y, Wang S, et al. Cytoprotective role of Ca2+-activated K+ channels in the cardiac inner
mitochondrial membrane. Science 2002;298:1029–33. [PubMed: 12411707]
54. Hausenloy D, Wynne A, Duchen M, Yellon D. Transient mitochondrial permeability transition pore
opening mediates preconditioning-induced protection. Circulation 2004;109:1714–7. [PubMed:
15066952]
Seubert et al. Page 9
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
55. Hausenloy DJ, Duchen MR, Yellon DM. Inhibiting mitochondrial permeability transition pore
opening at reperfusion protects against ischaemia-reperfusion injury. Cardiovasc Res 2003;60:617–
25. [PubMed: 14659807]
56. Weiss JN, Korge P, Honda HM, Ping P. Role of the mitochondrial permeability transition in
myocardial disease. Circ Res 2003;93:292–301. [PubMed: 12933700]
57. Wickenden A. K(+) channels as therapeutic drug targets. Pharmacol Ther 2002;94:157–82. [PubMed:
12191600]
58. Hu H, Sato T, Seharaseyon J, et al. Pharmacological and histochemical distinctions between
molecularly defined sarcolemmal kATP channels and native cardiac mitochondrial kATP channels.
Mol Pharmacol 1999;55:1000–5. [PubMed: 10347240]
59. Gross GJ, Peart JN. KATP channels and myocardial preconditioning: an update. Am J Physiol Heart
Circ Physiol 2003;285:H921–30. [PubMed: 12915383]
60. Gross GJ, Fryer RM. Sarcolemmal versus mitochondrial ATP-sensitive K+ channels and myocardial
preconditioning. Circ Res 1999;84:973–9. [PubMed: 10325234]
61. Lu T, Hoshi T, Weintraub NL, Spector AA, Lee HC. Activation of ATP-sensitive k(+) channels by
epoxyeicosatrienoic acids in rat cardiac ventricular myocytes. J Physiol 2001;537:811–27. [PubMed:
11744757]
62. Ardehali H, Chen Z, Ko Y, Mejia-Alvarez R, Marban E. Multiprotein complex containing succinate
dehydrogenase confers mitochondrial ATP-sensitive K+ channel activity. Proc Natl Acad Sci USA
2004;101:11880–5. [PubMed: 15284438]
63. Liu Y, Sato T, O'Rourke B, Marban E. Mitochondrial ATP-dependent potassium channels: novel
effectors of cardioprotection? Circulation 1998;97:2463–9. [PubMed: 9641699]
64. O'Rourke B. Evidence for mitochondrial K+ channels and their role in cardioprotection. Circ Res
2004;94:420–32. [PubMed: 15001541]
65. Brayden JE, Nelson MT. Regulation of arterial tone by activation of calcium-dependent potassium
channels. Science 1992;256:532–5. [PubMed: 1373909]
66. Calderone V. Large-conductance, ca(2+)-activated k(+) channels: function, pharmacology and drugs.
Curr Med Chem 2002;9:1385–95. [PubMed: 12132994]
67. Campbell WB, Harder DR. Endothelium-derived hyperpolarizing factors and vascular cytochrome
p450 metabolites of arachidonic acid in the regulation of tone. Circ Res 1999;84:484–8. [PubMed:
10066684]
68. Roman RJ. p-450 metabolites of arachidonic acid in the control of cardiovascular function. Physiol
Rev 2002;82:131–85. [PubMed: 11773611]
69. Kroetz DL, Xu F. Regulation and inhibition of arachidonic acid omega-hydroxylases and 20-hete
formation. Annu Rev Pharmacol Toxicol 2005;45:413–38. [PubMed: 15822183]
70. Siemen D, Loupatatzis C, Borecky J, Gulbins E, Lang F. Ca2+-activated k channel of the bk-type in
the inner mitochondrial membrane of a human glioma cell line. Biochem Biophys Res Commun
1999;257:549–54. [PubMed: 10198249]
71. Halestrap AP. Mitochondrial permeability: dual role for the ADP/ATP translocator? Nature
2004;430:983–4. [PubMed: 15329711]
72. Crompton M. The mitochondrial permeability transition pore and its role in cell death. Biochem J
1999;341(Pt 2):233–49. [PubMed: 10393078]
73. Nazareth W, Yafei N, Crompton M. Inhibition of anoxia-induced injury in heart myocytes by
cyclosporin a. J Mol Cell Cardiol 1991;23:1351–4. [PubMed: 1811053]
74. Griffiths EJ, Halestrap AP. Protection by cyclosporin a of ischemia/reperfusion-induced damage in
isolated rat hearts. J Mol Cell Cardiol 1993;25:1461–9. [PubMed: 7512654]
75. Griffiths EJ, Ocampo CJ, Savage JS, Stern MD, Silverman HS. Protective effects of low and high
doses of cyclosporin a against reoxygenation injury in isolated rat cardiomyocytes are associated
with differential effects on mitochondrial calcium levels. Cell Calcium 2000;27:87–95. [PubMed:
10756975]
76. Juhaszova M, Zorov DB, Kim SH, et al. Glycogen synthase kinase-3beta mediates convergence of
protection signaling to inhibit the mitochondrial permeability transition pore. J Clin Invest
2004;113:1535–49. [PubMed: 15173880]
Seubert et al. Page 10
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
77. Moffat MP, Ward CA, Bend JR, Mock T, Farhangkhoee P, Karmazyn M. Effects of
epoxyeicosatrienoic acids on isolated hearts and ventricular myocytes. Am J Physiol
1993;264:H1154–60. [PubMed: 8476093]
78. Seubert JM, Goralski K, Sinal CJ, Murphy E, Zeldin DC. Role of soluble epoxide hydrolase in
postischemic recovery of heart contractile function. Circulation 2004;110(Suppl):808.
79. Liu GS, Cohen MV, Mochly-Rosen D, Downey JM. Protein kinase c-epsilon is responsible for the
protection of preconditioning in rabbit cardiomyocytes. J Mol Cell Cardiol 1999;31:1937–48.
[PubMed: 10525430]
80. Cross HR, Murphy E, Bolli R, Ping P, Steenbergen C. Expression of activated pkc epsilon (pkc
epsilon) protects the ischemic heart, without attenuating ischemic h(+) production. J Mol Cell Cardiol
2002;34:361–7. [PubMed: 11945027]
81. Sato T, Saito T, Saegusa N, Nakaya H. Mitochondrial Ca2+-activated K+ channels in cardiac
myocytes: a mechanism of the cardioprotective effect and modulation by protein kinase a. Circulation
2005;111:198–203. [PubMed: 15623543]
82. Zeldin DC. Epoxygenase pathways of arachidonic acid metabolism. J Biol Chem 2001;276:36059–
62. [PubMed: 11451964]
83. Wu S, Moomaw CR, Tomer KB, Falck JR, Zeldin DC. Molecular cloning and expression of cyp2j2,
a human cytochrome p450 arachidonic acid epoxygenase highly expressed in heart. J Biol Chem
1996;271:3460–8. [PubMed: 8631948]
84. Lee CR, Bray NK, Fornage MS, et al. Genetic variation in soluble epoxide hydrolase (EPHX2) and
risk of coronary heart disease: the atherosclerosis risk in communities (ARIC) study. Hum Mol Genet.
2006[Epub]
Seubert et al. Page 11
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 1.
CYP epoxygenase and hydroxylase mediated metabolism of arachidonic acid.
Seubert et al. Page 12
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 2.
Schematic of proposed mechanisms of EET-mediated cardioprotection. CYP2J2-derived
eicosanoids, readily removed by sEH, activate K+ channels, p42/p44-MAPK, PKA and/or PKC
pathways leading to cardioprotection.
Seubert et al. Page 13
Prostaglandins Other Lipid Mediat. Author manuscript; available in PMC 2007 November 14.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... Myocardial infarction studies in rodents have provided additional mechanistic evidence for EETs and other epoxylipids to cardiovascular diseases [15,16,38]. EETs have been demonstrated to improve cardiac mitochondrial function, decrease inflammation, and oppose apoptosis to reduce cardiac fibrosis and hypertrophy (Fig. 2) [14,38]. ...
... Myocardial infarction studies in rodents have provided additional mechanistic evidence for EETs and other epoxylipids to cardiovascular diseases [15,16,38]. EETs have been demonstrated to improve cardiac mitochondrial function, decrease inflammation, and oppose apoptosis to reduce cardiac fibrosis and hypertrophy (Fig. 2) [14,38]. Cardiomyocyte mitochondrial function is regulated by EETs which improve heart function following ischemia [39,40,41]. ...
... Cardiomyocyte mitochondrial function is regulated by EETs which improve heart function following ischemia [39,40,41]. Cardioprotective mitochondrial actions were found in mice overexpressing the CYP2J2 epoxygenase or lacking sEH [38,39,41]. EET actions on vascular smooth muscle cell K + channels vasodilate coronary arteries and EET actions on cardiomyocyte K + channels oppose metabolic stress to decrease heart damage following ischemia [42]. ...
Article
Cardiovascular and heart diseases are leading causes of morbidity and mortality. Coronary artery endothelial and vascular dysfunction, inflammation, and mitochondrial dysfunction contribute to progression of heart diseases such as arrhythmias, congestive heart failure, and heart attacks. Classes of fatty acid epoxylipids and their enzymatic regulation by soluble epoxide hydrolase (sEH) have been implicated in coronary artery dysfunction, inflammation, and mitochondrial dysfunction in heart diseases. Likewise, genetic and pharmacological manipulations of epoxylipids have been demonstrated to have therapeutic benefits for heart diseases. Increasing epoxylipids reduce cardiac hypertrophy and fibrosis and improve cardiac function. Beneficial actions for epoxylipids have been demonstrated in cardiac ischemia reperfusion injury, electrical conductance abnormalities and arrhythmias, and ventricular tachycardia. This review discusses past and recent findings on the contribution of epoxylipids in heart diseases and the potential for their manipulation to treat heart attacks, arrhythmias, ventricular tachycardia, and heart failure.
... Our current findings suggest increased CYP2J2 and EpFA levels potentially represent a compensatory endogenous mechanism in attempt to protect the failing DCM heart. Upon formation from their N−3 and N−6 PUFA precursors, EpFAs are re-esterified into the plasma membrane of the cell (37). Cellular stress and injury can prompt the rapid release of membrane-bound EpFAs via phospholipase A 2 , making free EpFAs widely available for cellular use (37,38). ...
... Upon formation from their N−3 and N−6 PUFA precursors, EpFAs are re-esterified into the plasma membrane of the cell (37). Cellular stress and injury can prompt the rapid release of membrane-bound EpFAs via phospholipase A 2 , making free EpFAs widely available for cellular use (37,38). However, liberation also predisposes EpFAs to metabolism and inactivation by intracellular epoxide hydrolase enzymes. ...
Article
Full-text available
Objective Metabolites derived from N −3 and N −6 polyunsaturated fatty acids (PUFAs) have both beneficial and detrimental effects on the heart. However, contribution of these lipid mediators to dilated cardiomyopathy (DCM)-associated mitochondrial dysfunction remains unknown. This study aimed to characterize DCM-specific alterations in the PUFA metabolome in conjunction with cardiac mitochondrial quality in human explanted heart tissues. Methods Left ventricular tissues obtained from non-failing control (NFC) or DCM explanted hearts, were assessed for N −3 and N −6 PUFA metabolite levels using LC-MS/MS. mRNA and protein expression of CYP2J2, CYP2C8 and epoxide hydrolase enzymes involved in N −3 and N −6 PUFA metabolism were quantified. Cardiac mitochondrial quality was assessed by transmission electron microscopy, measurement of respiratory chain complex activities and oxygen consumption (respiratory control ratio, RCR) during ADP-stimulated ATP production. Results Formation of cardioprotective CYP-derived lipid mediators, epoxy fatty acids (EpFAs), and their corresponding diols were enhanced in DCM hearts. These findings were corroborated by increased expression of CYP2J2 and CYP2C8 enzymes, as well as microsomal and soluble epoxide hydrolase enzymes, suggesting enhanced metabolic flux and EpFA substrate turnover. DCM hearts demonstrated marked damage to mitochondrial ultrastructure and attenuated mitochondrial function. Incubation of fresh DCM cardiac fibers with the protective EpFA, 19,20-EDP, significantly improved mitochondrial function. Conclusions The current study demonstrates that increased expressions of CYP-epoxygenase enzymes and epoxide hydrolases in the DCM heart correspond with enhanced PUFA-derived EpFA turnover. This is accompanied by severe mitochondrial functional impairment which can be rescued by the administration of exogenous EpFAs.
... In nonischemic conditions, the mPTP remains closed, which maintains mitochondrial membrane potential. mPTP opening in response to reperfusion-induced oxidative stress results in the leakage of large molecules, loss of membrane potential, uncoupling of oxidative phosphorylation, and ultimately leads to cell death (18). Hearts with elevated EETs have increased activation of PI3 kinase, AKT, and/or ERK during early reperfusion (6,19). ...
Article
Full-text available
Cytochromes P450 (CYP) metabolize arachidonic acid (AA) to epoxyeicosatrienoic acids (EETs) which have numerous effects. After cardiac ischemia, EET-induced coronary vasodilation increases delivery of oxygen/nutrients to the myocardium, and EET-induced signaling protects cardiomyocytes against post-ischemic mitochondrial damage. Soluble epoxide hydrolase 2 (EPHX2) diminishes the benefits of EETs through hydrolysis to less active dihydroxyeicosatrienoic acids (DHETs). EPHX2 inhibition or genetic disruption improves recovery of cardiac function after ischemia. Immunohistochemical staining revealed EPHX2 expression in cardiomyocytes and some endothelial cells, but little expression in cardiac smooth muscle cells or fibroblasts. To determine specific roles of EPHX2 in cardiac cell types, we generated mice with cell-specific disruption of Ephx2 in endothelial cells (Ephx2fx/fx/Tek-cre) or cardiomyocytes (Ephx2fx/fx/Myh6-cre) to compare to global Ephx2-deficient mice (global Ephx2-/-) and wild-type (Ephx2fx/fx) mice in expression, EET hydrolase activity, and heart function studies. Most cardiac EPHX2 expression and activity is in cardiomyocytes with substantially less in endothelial cells. Ephx2fx/fx/Tek-cre hearts have similar EPHX2 expression, hydrolase activity, and post-ischemic cardiac function as control Ephx2fx/fx hearts. However, Ephx2fx/fx/Myh6-cre hearts were similar to global Ephx2-/- hearts with significantly diminished EPHX2 expression, decreased hydrolase activity, and enhanced post-ischemic cardiac function compared to Ephx2fx/fx hearts. During reperfusion, Ephx2fx/fx/Myh6-cre hearts displayed increased ERK activation compared to Ephx2fx/fx hearts which could be reversed by EEZE treatment. EPHX2 did not regulate coronary vasodilation in this model. We conclude that EPHX2 is primarily expressed in cardiomyocytes where it regulates EET hydrolysis and post-ischemic cardiac function, whereas endothelial EPHX2 does not play a significant role in these processes.
... GSK1016790A (a TRPV4 agonist) dose-dependently induced hippocampal neuronal death, along with increased phosphorylation of the NR2B subunit of the N-methyl-D-aspartate receptor (NMDAR) (Jie et al., 2016). TRPV4 is also activated by arachidonic acid, cell swelling, or epoxyeicosatrienoic acids in neurons, which are associated with cerebral ischemia (Seubert et al.., 2007). Activation of the TRPV4 channel upregulated Ca 2+ entry via NMDARs and required the NR2B subunit . ...
Article
Full-text available
Methods The databases Pubmed, and the National Library of Medicine were searched for literature. All papers on celebral stroke and transient receptor potential ion channels were considered. Results Stroke is the second leading cause of death and disability, with an increasing incidence in developing countries. About 75 per cent of strokes are caused by occlusion of cerebral arteries, and substantial advances have been made in elucidating mechanisms how stroke affects the brain. Transient receptor potential (TRP) ion channels are calcium‐permeable channels highly expressed in brain that drives Ca²⁺ entry into multiple cellular compartments. TRPC1/3/4/6, TRPV1/2/4, and TRPM2/4/7 channels have been implicated in stroke pathophysiology. Conclusions Although the precise mechanism of transient receptor potential ion channels in cerebral stroke is still unclear, it has the potential to be a therapeutic target for patients with stroke if developed appropriately. Hence, more research is needed to prove its efficacy in this context.
... EETs promote the growth of a primary tumor and metastasis as well as the exit of a tumor from an indolent state [309]. The functions of EETs are associated with carcinogenesis, metastasis of various cancers (including colon, liver, kidney, breast, and prostate cancers [309][310][311][312][313]), with angiogenesis [314][315][316][317], and restoration of cardiac tissue after ischemic stroke [318]. ...
Article
Full-text available
CYP3A is an enzyme subfamily in the cytochrome P450 (CYP) superfamily and includes isoforms CYP3A4, CYP3A5, CYP3A7, and CYP3A43. CYP3A enzymes are indiscriminate toward substrates and are unique in that these enzymes metabolize both endogenous compounds and diverse xenobiotics (including drugs); almost the only common characteristic of these compounds is lipophilicity and a relatively large molecular weight. CYP3A enzymes are widely expressed in human organs and tissues, and consequences of these enzymes’ activities play a major role both in normal regulation of physiological levels of endogenous compounds and in various pathological conditions. This review addresses these aspects of regulation of CYP3A enzymes under physiological conditions and their involvement in the initiation and progression of diseases.
... On the other hand, human CYP2C9 and CYP2J2 convert ARA to epoxyeicosatrienoic acids (EETs) via epoxygenation. These EETs have anti-inflammatory properties, inhibit platelet aggregation [14], have a vasodilatory effect, and prevent hypertension-induced renal damage [15,16]. Under conditions of normal body homeostasis, CYP hydroxylation and epoxygenation remain in balance. ...
Article
Full-text available
Background An imbalance in the levels of arachidonic acid (ARA) metabolites in cardiovascular disorders and drug-induced cardiotoxicity have been previously described. Aims This study aimed to investigate the influence of cyclooxygenase-2 (COX-2) selective inhibitors on the gene expression of ARA-metabolizing genes and beta1 gene in the hearts and kidneys of experimental mice. Methods Thirty-five balb/c mice were divided into five groups with seven mice per group. The groups were then given two distinct types of COX-2 selective inhibitors, rofecoxib and celecoxib, in two different doses equivalent to those used in human treatment for 30 days. The mRNA expression of beta1, ace2, and ARA-metabolizing genes, coxs, lipoxygenases (aloxs), and cytochrome p450 (cyp450s) in mice heart and kidneys were assessed. Genes were analyzed using real-time polymerase chain reaction analysis. In addition, rofecoxib-induced histological alterations were examined. Results It was found that only the high dose of rofecoxib (5 mg/kg) caused toxicological alterations, a finding that was indicated by a significant increase (P < 0.05) in the relative weight of the mouse hearts and increase in the ventricle wall thickness as observed through pathohistological examination. This increase was associated with a significant increase in the mRNA expression level of the beta1 receptor in both the heart and kidneys of the mice (53- and 12-fold, respectively). The expression of both cox1 and 2 genes was increased 4-fold in the kidneys. In addition, the expression of alox12 gene increased significantly (by 67-fold in the heart and by 21-fold in the kidney), while alox15 gene expression was upregulated in the heart by 8-fold and 5-fold in the kidney. The genes responsible for synthesizing 20-Hydroxyeicosatetraenoic acid (cyp4a12 and cyp1a1) were significantly upregulated (P < 0.05) in the hearts of high-dose rofecoxib-treated mice by 7- and 17 -fold, respectively. In addition, the expression of epoxyeicosatrienoic acid-synthesizing genes, cyp2c29 and cyp2j5, was increased significantly (P < 0.05) in the hearts of high-dose rofecoxib-treated mice by 4- and 16-fold, respectively. Conclusion Rofecoxib caused upregulation of the mRNA expression of beta 1 gene in association with increased expression of ARA-metabolizing genes in mouse hearts and kidneys. These findings may help us understand the molecular cardiotoxic mechanism of rofecoxib.
... The CYP450 pathway, includes epoxygenase and ω-hydroxylase, which generate epoxyeicosatrienoic acids (EETs) and 20-hydroxy-eicosatetraenoic acids (20-HETE), respectively. These metabolites also play a role in the regulation of vascular injury [25,26]. The 20-HETE is a pro-inflammatory metabolite with potent vasoconstrictor properties that promotes endothelial dysfunction and progression of CVD [27], whereas EETs are endothelial derived hyperpolarizing factors with anti-inflammatory properties and beneficial effects in CVD28,29. ...
Article
Full-text available
Hyperhomocysteinemia (HHcy) contributes to the incidence of many cardiovascular diseases (CVD). Our group have previously established crucial roles of eicosanoids and homocysteine in the incidence of vascular injury in diabetic retinopathy and renal injury. Using cystathionine-β-synthase heterozygous mice (cβs+/-) as a model of HHcy, the current study was designed to determine the impact of homocysteine on circulating levels of lipid mediators derived from polyunsaturated fatty acids (PUFA). Plasma samples were isolated from wild-type (WT) and cβs+/- mice for the assessment of eicosanoids levels using LC/MS. Plasma 12/15-lipoxygenase (12/15-LOX) activity significantly decreased in cβs+/- vs. WT control mice. LOX-derived metabolites from both omega-3 and omega-6 PUFA were also reduced in cβs+/- mice compared to WT control (P < 0.05). Contrary to LOX metabolites, cytochrome P450 (CYP) metabolites from omega-3 and omega-6 PUFA were significantly elevated in cβs+/- mice compared to WT control. Epoxyeicosatrienoic acids (EETs) are epoxides derived from arachidonic acid (AA) metabolism by CYP with anti-inflammatory properties and are known to limit vascular injury, however their physiological role is limited by their rapid degradation by soluble epoxide hydrolase (sEH) to their corresponding diols (DiHETrEs). In cβs+/- mice, a significant decrease in the plasma EETs bioavailability was obvious as evident by the decrease in EETs/ DiHETrEs ratio relative to WT control mice. Cyclooxygenase (COX) metabolites were also significantly decreased in cβs+/- vs. WT control mice. These data suggest that HHcy impacts eicosanoids metabolism through decreasing LOX and COX metabolic activities while increasing CYP metabolic activity. The increase in AA metabolism by CYP was also associated with increase in sEH activity and decrease in EETs bioavailability. Dysregulation of eicosanoids metabolism could be a contributing factor to the incidence and progression of HHcy-induced CVD.
Article
Deterioration of physiological systems, like the cardiovascular system, occurs progressively with age impacting an individual's health and increasing susceptibility to injury and disease. Cellular senescence has an underlying role in age-related alterations and can be triggered by natural aging or prematurely by stressors such as the bacterial toxin, lipopolysaccharide (LPS). The metabolism of polyunsaturated fatty acids (PUFAs) by CYP450 enzymes produces numerous bioactive lipid mediators that can be further metabolized by soluble epoxide hydrolase (sEH) into diol metabolites, often with reduced biological effects. In our study, we observed age-related cardiac differences in female mice, where young mice demonstrated resistance to LPS injury, and genetic deletion or pharmacological inhibition of sEH using trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid ( tAUCB) attenuated LPS-induced cardiac dysfunction in aged female mice. Bulk RNA-sequencing analyses revealed transcriptomics differences in aged female hearts. Confirmatory analysis demonstrated changes to inflammatory and senescence genes markers such as Il-6, Mcp1, Il-1β, Nlrp3, p21, p16, SA-β-gal, and Gdf15 were attenuated in the hearts of aged female mice where sEH was deleted or inhibited. Collectively, these findings highlight the role of sEH in modulating the aging process of the heart, whereby targeting sEH is cardioprotective.
Chapter
Drug-metabolizing enzymes (DMEs) belong to a multigene superfamily of enzymes, which are essential for the metabolism of many endogenous and foreign substances. Among DMEs, cytochrome P450 (CYP450) enzyme system, and its various isoenzymes are among the major components of mixed function oxidases enzymes. Various pathophysiological factors, genetic variations, and drug interactions can result in decreased, absent, and/or enhanced activity of DMEs. The alterations in the activity of CYPs greatly affect the response of an individual against the therapeutic treatment. Also, there are limited data about the influence caused by these changes on the CYPs functional roles in the pathophysiological and physiological actions of the cardiovascular system. Many tissues other than liver, such as heart, possess active CYP450 enzymes, but there is limited information related to their influence in homeostasis or cellular injury. This chapter tries to explore the potential therapeutic targets in the treatment of major cardiovascular conditions where genotyping of DMEs that are genetically polymorphic may be considered as beneficial for therapeutic outcome or drug safety. This chapter investigates the expression and localization of phase I and phase II DMEs in the heart regulated via AhR pathway in various species along with brief demonstration of their protein and mRNA activity both at the inducible and basal levels. It also demonstrates the pharmacogenomics of cardiovascular diseases and drugs and their association with DMEs along with the gender specific variation of DMEs in the pharmacology of cardiovascular events.
Article
Full-text available
A cDNA encoding a human cytochrome P450 arachidonic acid epoxygenase was isolated from a human liver cDNA library. Sequence analysis revealed that this 1,876-base pair cDNA contained an open reading frame and encoded a new 502-amino acid protein designated CYP2J2. Blot hybridization analysis of RNA prepared from human tissues revealed that CYP2J2 was highly expressed in the heart. Recombinant CYP2J2 protein was prepared using the baculovirus expression system and purified to near electrophoretic homogeneity. The enzyme metabolized arachidonic acid predominantly via olefin epoxidation to all four regioisomeric cis-epoxyeicosatrienoic acids (catalytic turnover 65 pmol of product formed/nmol of cytochrome P450/min at 30°C). Epoxidation of arachidonic acid by CYP2J2 at the 14,15-olefin was highly enantioselective for (14R,15S)-epoxyeicosatrienoic acid (76% optical purity). Immunoblotting of microsomal fractions prepared from human tissues using a polyclonal antibody raised against the recombinant hemoprotein confirmed primary expression of CYP2J2 protein in human heart. The in vivo significance of CYP2J2 was suggested by documenting the presence of epoxyeicosatrienoic acids in the human heart using gas chromatography/mass spectroscopy. Importantly, the chirality of CYP2J2 products matched that of the epoxyeicosatrienoic acid enantiomers present, in vivo, in human heart. We propose that CYP2J2 is one of the enzymes responsible for epoxidation of endogenous arachidonic acid pools in human heart and that epoxyeicosatrienoic acids may, therefore, play important functional roles in cardiac physiology.
Article
A variety of direct and indirect techniques have revealed the existence of ATP-sensitive potassium (K-ATP) channels in the inner membranes of mitochondria. The molecular identity of these mitochondrial K-ATP, (mitoK(ATP)) channels remains unclear. We used a pharmacological approach to distinguish mitoK(ATP) channels from classical, molecularly defined cardiac sarcolemmal K-ATP (surfaceK(ATP)) channels encoded by the sulfonylurea receptor SUR2A and the pore-forming subunit K(lr)6.2. SUR2A and K(ir)6.2 were expressed in human embryonic kidney (HEK)293 cells, and their activities were measured by patch-clamp recordings of membrane current. SurfaceK(ATP) channels are activated potently by 100 mu M pinacidil but only weakly by 100 mu M diazoxide; in addition, they are blocked by 10 mu M glibenclamide, but are insensitive to 500 mu M 5-hydroxydecanoate. This pharmacology, which was confirmed with patch-clamp recordings in intact rabbit: ventricular myocytes, contrasts with that of mitoK(ATP) channels as indexed by flavoprotein oxidation. MitoK(ATP) channels in myocytes are activated equally by 100 mu M diazoxide and 100 mu M pinacidil. In contrast to its lack of effect on surfaceK(ATP) channels, 5-hydroxydecanoate is an effective blocker of mitoK(ATP) channels. Glibenclamide's effects on mitoK(ATP) channels are difficult to assess, because it independently activates flavoprotein fluorescence, consistent with a previously described primary uncoupling effect. Confocal imaging of the subcellular distribution of expressed fluorescent K(ir)6.2 in HEK cells and in myocytes revealed no targeting of mitochondrial membranes. The differences in drug sensitivity and subcellular localization indicate that mitoK(ATP) channels are distinct from surface K-ATP channels at a molecular level.
Article
This article reviews the involvement of the mitochondrial permeability transition pore in necrotic and apoptotic cell death. The pore is formed from a complex of the voltage-dependent anion channel (VDAC), the adenine nucleotide translocase and cyclophilin-D (CyP-D) at contact sites between the mitochondrial outer and inner membranes. In vitro, under pseudopathological conditions of oxidative stress, relatively high Ca2+ and low ATP, the complex flickers into an open-pore state allowing free diffusion of low-Mr solutes across the inner membrane. These conditions correspond to those that unfold during tissue ischaemia and reperfusion, suggesting that pore opening may be an important factor in the pathogenesis of necrotic cell death following ischaemia/reperfusion. Evidence that the pore does open during ischaemia/reperfusion is discussed. There are also strong indications that the VDAC-adenine nucleotide translocase-CyP-D complex can recruit a number of other proteins, including Bax, and that the complex is utilized in some capacity during apoptosis. The apoptotic pathway is amplified by the release of apoptogenic proteins from the mitochondrial intermembrane space, including cytochrome c, apoptosis-inducing factor and some procaspases. Current evidence that the pore complex is involved in outer-membrane rupture and release of these proteins during programmed cell death is reviewed, along with indications that transient pore opening may provoke 'accidental' apoptosis.
Article
A cDNA encoding a P450 monooxygenase was amplified from reverse transcribed rat heart and liver total RNA by polymerase chain reaction using primers based on the 5′- and 3′-end sequences of two rat pseudogenes, CYP2J3P1 andCYP2J3P2. Sequence analysis revealed that this 1,778-base pair cDNA contained an open reading frame and encoded a new 502 amino acid protein designated CYP2J3. Based on the deduced amino acid sequence, CYP2J3 was approximately 70% homologous to both human CYP2J2 and rabbit CYP2J1. Recombinant CYP2J3 protein was co-expressed with NADPH-cytochrome P450 oxidoreductase in Sf9 insect cells using a baculovirus expression system. Microsomal fractions of CYP2J3/NADPH-cytochrome P450 oxidoreductase-transfected cells metabolized arachidonic acid to 14,15-, 11,12-, and 8,9-epoxyeicosatrienoic acids and 19-hydroxyeicosatetraenoic acid as the principal reaction products (catalytic turnover, 0.2 nmol of product/nmol of cytochrome P450/min at 37 °C). Immunoblotting of microsomal fractions prepared from rat tissues using a polyclonal antibody raised against recombinant CYP2J2 that cross-reacted with CYP2J3 but not with other known rat P450s demonstrated abundant expression of CYP2J3 protein in heart and liver. Immunohistochemical staining of formalin-fixed paraffin-embedded rat heart tissue sections using the anti-CYP2J2 IgG and avidin-biotin-peroxidase detection localized expression of CYP2J3 primarily to atrial and ventricular myocytes. In an isolated-perfused rat heart model, 20 min of global ischemia followed by 40 min of reflow resulted in recovery of only 44 ± 6% of base-line contractile function. The addition of 5 μm 11,12-epoxyeicosatrienoic acid to the perfusate prior to global ischemia resulted in a significant 1.6-fold improvement in recovery of cardiac contractility (69 ± 5% of base line,p = 0.01 versus vehicle alone). Importantly, neither 14,15-epoxyeicosatrienoic acid nor 19-hydroxyeicosatetraenoic acid significantly improved functional recovery following global ischemia, demonstrating the specificity of the biological effect for the 11,12-epoxyeicosatrienoic acid regioisomer. Based on these data, we conclude that (a) CYP2J3 is one of the predominant enzymes responsible for the oxidation of endogenous arachidonic acid pools in rat heart myocytes and (b) 11,12-epoxyeicosatrienoic acid may play an important functional role in the response of the heart to ischemia.
Article
: Pharmacological evidence has implicated ATP-dependent potassium (KATP) channels in the mechanism of ischemic preconditioning; however, the effects of sarcolemmal KATP channels on excitability cannot account for the protection. KATP channels also exist in mitochondrial inner membrane. To test whether such channels play a role in cardioprotection, we simultaneously measured flavoprotein fluorescence, an index of mitochondrial redox state, and sarcolemmal KATP currents in intact rabbit ventricular myocytes. Our results show that diazoxide, a KATP channel opener, induced reversible oxidation of flavoproteins, but did not activate sarcolemmal KATP channels. This effect of diazoxide was blocked by 5-hydroxydecanoic acid (5-HD). We further verified that 5-HD is a selective blocker of the mitochondrial KATP channels. These methods have enabled us to demonstrate that the activity of mitochondrial KATP channels can be regulated by protein kinase C. In a cellular model of simulated ischemia, inclusion of diazoxide decreased the rate of cell death to about half of that in control. Such protection is inhibited by 5-HD. In conclusion, our results demonstrate that diazoxide targets mitochondrial but not sarcolemmal KATP channels, and imply that mitochondrial KATP channels may mediate pre-conditioning.
Article
A cDNA encoding a P450 monooxygenase was amplified from reverse transcribed rat heart and liver total RNA by polymerase chain reaction using primers based on the 5′- and 3′-end sequences of two rat pseudogenes, CYP2J3P1 andCYP2J3P2. Sequence analysis revealed that this 1,778-base pair cDNA contained an open reading frame and encoded a new 502 amino acid protein designated CYP2J3. Based on the deduced amino acid sequence, CYP2J3 was approximately 70% homologous to both human CYP2J2 and rabbit CYP2J1. Recombinant CYP2J3 protein was co-expressed with NADPH-cytochrome P450 oxidoreductase in Sf9 insect cells using a baculovirus expression system. Microsomal fractions of CYP2J3/NADPH-cytochrome P450 oxidoreductase-transfected cells metabolized arachidonic acid to 14,15-, 11,12-, and 8,9-epoxyeicosatrienoic acids and 19-hydroxyeicosatetraenoic acid as the principal reaction products (catalytic turnover, 0.2 nmol of product/nmol of cytochrome P450/min at 37 °C). Immunoblotting of microsomal fractions prepared from rat tissues using a polyclonal antibody raised against recombinant CYP2J2 that cross-reacted with CYP2J3 but not with other known rat P450s demonstrated abundant expression of CYP2J3 protein in heart and liver. Immunohistochemical staining of formalin-fixed paraffin-embedded rat heart tissue sections using the anti-CYP2J2 IgG and avidin-biotin-peroxidase detection localized expression of CYP2J3 primarily to atrial and ventricular myocytes. In an isolated-perfused rat heart model, 20 min of global ischemia followed by 40 min of reflow resulted in recovery of only 44 ± 6% of base-line contractile function. The addition of 5 μm 11,12-epoxyeicosatrienoic acid to the perfusate prior to global ischemia resulted in a significant 1.6-fold improvement in recovery of cardiac contractility (69 ± 5% of base line,p = 0.01 versus vehicle alone). Importantly, neither 14,15-epoxyeicosatrienoic acid nor 19-hydroxyeicosatetraenoic acid significantly improved functional recovery following global ischemia, demonstrating the specificity of the biological effect for the 11,12-epoxyeicosatrienoic acid regioisomer. Based on these data, we conclude that (a) CYP2J3 is one of the predominant enzymes responsible for the oxidation of endogenous arachidonic acid pools in rat heart myocytes and (b) 11,12-epoxyeicosatrienoic acid may play an important functional role in the response of the heart to ischemia.
Article
1. Arachidonic acid (AA) is metabolized by cytochrome P450 (CYP)-dependent pathways to epoxyeicosatrienoic acids (EET) and 20-hydroxyeicosatetraenoic acid (20-HETE) in the kidney and the peripheral vasculature. 2. The present short review summarizes the renal and cardiovascular actions of these important mediators. 3. Epoxyeicosatrienoic acids are vasodilators produced by the endothelium that hyperpolarize vascular smooth muscle (VSM) cells by opening Ca2+-activated K+ (KCa) channels. 20-Hydroxyeicosatetraenoic acid is a vasoconstrictor that inhibits the opening of KCa channels in VSM cells. Cytochrome P450 4A inhibitors block the myogenic response of small arterioles to elevations in transmural pressure and autoregulation of renal and cerebral blood flow in vivo. Cytochrome P450 4A blockers also attenuate the vasoconstrictor response to elevations in tissue PO2, suggesting that this system may serve as a vascular oxygen sensor. Nitric oxide and carbon monoxide inhibit the formation of 20-HETE and a fall in 20-HETE levels contributes to the activation of KCa channels in VSM cells and the vasodilator response to these gaseous mediators. 20-Hydroxyeicosatetraenoic acid also mediates the inhibitory actions of peptide hormones on sodium transport in the kidney and the mitogenic effects of growth factors in VSM and mesangial cells. A deficiency in the renal production of 20-HETE is associated with the development of hypertension in Dahl salt-sensitive rats. 4. In summary, the available evidence indicates that CYP metabolites of AA play a central role in the regulation of renal, pulmonary and vascular function and that abnormalities in this system may contribute to the pathogenesis of cardiovascular diseases.